1
|
Jiang H, Gao J, Wang H, Zhao L, Yang Y, Ma J, Gu S, Hu F, Du Q, Wang F. Rehydration effect of qingshu buye decoction on exercise and high temperature-induced dehydration. J Int Soc Sports Nutr 2024; 21:2393364. [PMID: 39161283 PMCID: PMC11338198 DOI: 10.1080/15502783.2024.2393364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVE The aim of this study was to conduct a comprehensive evaluation of the rehydration efficacy of QSBYD and elucidate its potential underlying mechanism. DESIGN 38 participants were randomly assigned to receive either QSBYD or placebo before and after exercise and heat-induced dehydration. Hydration indicators were measured over time. Blood tests assessed cellular anaerobic respiration metabolites, serum inflammatory markers, and coagulation markers. Perceptual measures of thirst, fatigue, and muscular soreness were also taken. RESULTS QSBYD consumption resulted in lower urine volume (Control vs. QSBYD: 260.83 ± 167.99 ml vs. 187.78 ± 141.34 ml) and smaller decrease in percentage of nude body weight change from baseline (Control vs. QSBYD: -0.52 ± 0.89% vs. -0.07 ± 0.52%). Although no significant differences in urine specific gravity, QSBYD resulted in reduced urine volume at 120 min, suggesting improved fluid retention. Furthermore, QSBYD resulted in lower levels of IL-1β (Control vs. QSBYD: 2.40 ± 0.68 vs. 1.33 ± 0.66 pg/mL), suggesting QSBYD may provide benefits beyond hydration. CONCLUSION Further investigation into the underlying mechanisms and long-term effects of QSBYD on hydration is warranted. QSBYD may be an effective alternative to commercial sports drinks in mitigating dehydration effects.
Collapse
Affiliation(s)
- Huanyu Jiang
- Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu, China
| | - Jiankun Gao
- Sichuan College of Traditional Chinese Medicine, School of pharmacy, Mianyang, China
| | - Huan Wang
- Sichuan College of Traditional Chinese Medicine, Laboratory Training Center, Mianyang, China
| | - Lin Zhao
- Sichuan College of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Mianyang, China
| | - Yingduo Yang
- University of Southern California, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, USA
| | - Jiahua Ma
- Sichuan Depeiyuan Traditional Chinese Medicine Science & Technology Development Co., Ltd., Mianyang, China
- Sichuan Dekeyuan Science and Technology Development Co., Ltd., Mianyang, China
| | - Shan Gu
- Sichuan Academy of Safety Science and Technology, Research and Development Department, Chengdu, China
| | - Fenglin Hu
- Sichuan Academy of Safety Science and Technology, Research and Development Department, Chengdu, China
| | - Quanyu Du
- Hospital of Chengdu University of Traditional Chinese Medicine, Department of endocrinology, Chengdu, China
| | - Fei Wang
- Sichuan College of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
2
|
Packer M, Ferreira JP, Butler J, Filippatos G, Januzzi JL, González Maldonado S, Panova-Noeva M, Pocock SJ, Prochaska JH, Saadati M, Sattar N, Sumin M, Anker SD, Zannad F. Reaffirmation of Mechanistic Proteomic Signatures Accompanying SGLT2 Inhibition in Patients With Heart Failure: A Validation Cohort of the EMPEROR Program. J Am Coll Cardiol 2024:S0735-1097(24)07897-5. [PMID: 39217550 DOI: 10.1016/j.jacc.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors exert a distinctive pattern of direct biological effects on the heart and kidney under experimental conditions, but the meaningfulness of these signatures for patients with heart failure has not been fully defined. OBJECTIVES We performed the first mechanistic validation study of large-scale proteomics in a double-blind randomized trial of any treatment in patients with heart failure. METHODS In a discovery cohort from the EMPEROR (Empagliflozin Outcome Trial in Patients With Chronic Heart Failure and Reduced Ejection Fraction) program, we studied the effect of randomized treatment with placebo or empagliflozin on 1,283 circulating proteins in 1,134 patients with heart failure with a reduced or preserved ejection fraction. In a validation cohort, we expanded the number to 2,155 assessed proteins, which were measured in 1,120 EMPEROR participants who had not been studied previously. RESULTS In the validation cohort, 25 proteins were the most differentially enriched by empagliflozin (ie, ≥15% between-group difference and false discovery rate <1% at 12 weeks with known effects on the heart or kidney): 1) 13 proteins promote autophagy and other cellular quality-control functions (IGFBP1, OTUB1, DNAJB1, DNAJC9, RBP2, IST1, HSPA8, H-FABP, FABP6, ATPIFI, TfR1, EPO, IGBP1); 2) 12 proteins enhance mitochondrial health and ATP production (UMtCK, TBCA, L-FABP, H-FABP, FABP5, FABP6, RBP2, IST1, HSPA8, ATPIFI, TfR1, EPO); 3) 7 proteins augment cellular iron mobilization or erythropoiesis (TfR1, EPO, IGBP1, ERMAP, UROD, ATPIF1, SNCA); 4) 3 proteins influence renal tubular sodium handling; and 5) 9 proteins have restorative effects in the heart or kidneys, with many proteins exerting effects in >1 domain. These biological signatures replicated those observed in our discovery cohort. When the threshold for a meaningful between-group difference was lowered to ≥10%, there were 58 additional differentially enriched proteins with actions on the heart and kidney, but the biological signatures remained the same. CONCLUSIONS The replication of mechanistic signatures across discovery and validation cohorts closely aligns with the experimental effects of SGLT2 inhibitors. Thus, the actions of SGLT2 inhibitors-to promote autophagy, restore mitochondrial health and production of ATP, promote iron mobilization and erythropoiesis, influence renal tubular ion reabsorption, and normalize cardiac and renal structure and function-are likely to be relevant to patients with heart failure. (EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure With Preserved Ejection Fraction [EMPEROR-Preserved], NCT03057951; EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure With Reduced Ejection Fraction [EMPEROR-Reduced], NCT03057977).
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas, USA; Imperial College London, London, United Kingdom.
| | - João Pedro Ferreira
- UnIC@RISE, Cardiovascular Research and Development Center, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal; Baylor Scott and White Research Institute, Dallas, Texas, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, USA; University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens School of Medicine, Athens University Hospital Attikon, Athens, Greece
| | - James L Januzzi
- National and Kapodistrian University of Athens School of Medicine, Athens University Hospital Attikon, Athens, Greece; Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | | | - Marina Panova-Noeva
- Boehringer Ingelheim Pharma GmbH & Co KG, Ingelheim, Germany; Center for Thrombosis and Haemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stuart J Pocock
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jürgen H Prochaska
- Boehringer Ingelheim International GmbH, Ingelheim, Germany; Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maral Saadati
- Elderbrook Solutions GmbH, on behalf of Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité, Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France; F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre Hospitalier Régional Universitaire de Nancy, Nancy, France
| |
Collapse
|
3
|
Barr SI, Bessa SS, Mohamed TM, Abd El-Azeem EM. Exosomal UMOD gene expression and urinary uromodulin level as early noninvasive diagnostic biomarkers for diabetic nephropathy in type 2 diabetic patients. Diabetol Int 2024; 15:389-399. [PMID: 39101162 PMCID: PMC11291796 DOI: 10.1007/s13340-023-00686-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/11/2023] [Indexed: 08/06/2024]
Abstract
Background Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. Exosomes are promising biomarkers for disease diagnosis and uromodulin is a kidney-specific protein. So, this study was designed to investigate the change in the gene expression of urinary exosomal uromodulin mRNA and urinary uromodulin level and determine the diagnostic potential of these noninvasive biomarkers in the early stage of diabetic nephropathy in type 2 diabetic patients. Method This study included 100 participants; urinary exosomes were isolated using polyethylene glycol (PEG). Gene expression of exosomal uromodulin mRNA was determined by quantitative real-time polymerase chain reaction (q-RT-PCR). The urinary uromodulin levels were determined by an enzyme-linked immunosorbent assay (ELISA). Result In this study, the gene expression of exosomal uromodulin (UMOD) mRNA and the level of urinary uromodulin showed a significant increase in all diabetic groups with and without nephropathy compared to the control group. The exosomal UMOD mRNA showed a significant positive correlation with urinary uromodulin in all groups. Multiple logistic regression showed that urinary uromodulin was an independent determinant for DN. A diagnostic model of two indicators, exosomal UMOD mRNA and urinary uromodulin, can significantly predict DN. The area under the curve is 0.095, with a 95% confidence interval of 0.98-1, and 0.81, with a 95% confidence interval of 0.69-0.92, for the exosomal UMOD mRNA and urinary uromodulin, respectively. Conclusion Urinary exosomal mRNA of UMOD and urinary uromodulin levels are progressively elevated in an early stage of DN, even before the microalbuminuria stage, so they could be used as early predictors for DN.
Collapse
Affiliation(s)
- Shaimaa I. Barr
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Sahar S. Bessa
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Tarek M. Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | | |
Collapse
|
4
|
Mercado-Evans V, Chew C, Serchejian C, Saltzman A, Mejia ME, Zulk JJ, Cornax I, Nizet V, Patras KA. Tamm-Horsfall protein augments neutrophil NETosis during urinary tract infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578501. [PMID: 38370726 PMCID: PMC10871275 DOI: 10.1101/2024.02.01.578501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Urinary neutrophils are a hallmark of urinary tract infection (UTI), yet the mechanisms governing their activation, function, and efficacy in controlling infection remain incompletely understood. Tamm-Horsfall glycoprotein (THP), the most abundant protein in urine, uses terminal sialic acids to bind an inhibitory receptor and dampen neutrophil inflammatory responses. We hypothesized that neutrophil modulation is an integral part of THP-mediated host protection. In a UTI model, THP-deficient mice showed elevated urinary tract bacterial burdens, increased neutrophil recruitment, and more severe tissue histopathological changes compared to WT mice. Furthermore, THP-deficient mice displayed impaired urinary NETosis during UTI. To investigate the impact of THP on NETosis, we coupled in vitro fluorescence-based NET assays, proteomic analyses, and standard and imaging flow cytometry with peripheral human neutrophils. We found that THP increases proteins involved in respiratory chain, neutrophil granules, and chromatin remodeling pathways, enhances NETosis in an ROS-dependent manner, and drives NET-associated morphologic features including nuclear decondensation. These effects were observed only in the presence of a NETosis stimulus and could not be solely replicated with equivalent levels of sialic acid alone. We conclude that THP is a critical regulator of NETosis in the urinary tract, playing a key role in host defense against UTI.
Collapse
Affiliation(s)
- Vicki Mercado-Evans
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Claude Chew
- Cytometry and Cell Sorting Core, Baylor College of Medicine, Houston, Texas, USA
| | - Camille Serchejian
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Alexander Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Marlyd E. Mejia
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ingrid Cornax
- Department of Pediatrics, UC San Diego, La Jolla, California, USA
| | - Victor Nizet
- Department of Pediatrics, UC San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
5
|
Takata T, Isomoto H. The Versatile Role of Uromodulin in Renal Homeostasis and Its Relevance in Chronic Kidney Disease. Intern Med 2024; 63:17-23. [PMID: 36642527 PMCID: PMC10824655 DOI: 10.2169/internalmedicine.1342-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/06/2022] [Indexed: 01/15/2023] Open
Abstract
Uromodulin, also known as the Tamm-Horsfall protein, is predominantly expressed in epithelial cells of the kidney. It is secreted mainly in the urine, although small amounts are also found in serum. Uromodulin plays an important role in maintaining renal homeostasis, particularly in salt/water transport mechanisms and is associated with salt-sensitive hypertension. It also regulates urinary tract infections, kidney stones, and the immune response in the kidneys or extrarenal organs. Uromodulin has been shown to be associated with the renal function, age, nephron volume, and metabolic abnormalities and has been proposed as a novel biomarker for the tubular function or injury. These findings suggest that uromodulin is a key molecule underlying the mechanisms or therapeutic approaches of chronic kidney disease, particularly nephrosclerosis and diabetic nephropathy, which are causes of end-stage renal disease. This review focuses on the current understanding of the role of uromodulin from a biological, physiological, and pathological standpoint.
Collapse
Affiliation(s)
- Tomoaki Takata
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Japan
| |
Collapse
|
6
|
Thielemans R, Speeckaert R, Delrue C, De Bruyne S, Oyaert M, Speeckaert MM. Unveiling the Hidden Power of Uromodulin: A Promising Potential Biomarker for Kidney Diseases. Diagnostics (Basel) 2023; 13:3077. [PMID: 37835820 PMCID: PMC10572911 DOI: 10.3390/diagnostics13193077] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Uromodulin, also known as Tamm-Horsfall protein, represents the predominant urinary protein in healthy individuals. Over the years, studies have revealed compelling associations between urinary and serum concentrations of uromodulin and various parameters, encompassing kidney function, graft survival, cardiovascular disease, glucose metabolism, and overall mortality. Consequently, there has been a growing interest in uromodulin as a novel and effective biomarker with potential applications in diverse clinical settings. Reduced urinary uromodulin levels have been linked to an elevated risk of acute kidney injury (AKI) following cardiac surgery. In the context of chronic kidney disease (CKD) of different etiologies, urinary uromodulin levels tend to decrease significantly and are strongly correlated with variations in estimated glomerular filtration rate. The presence of uromodulin in the serum, attributable to basolateral epithelial cell leakage in the thick ascending limb, has been observed. This serum uromodulin level is closely associated with kidney function and histological severity, suggesting its potential as a biomarker capable of reflecting disease severity across a spectrum of kidney disorders. The UMOD gene has emerged as a prominent locus linked to kidney function parameters and CKD risk within the general population. Extensive research in multiple disciplines has underscored the biological significance of the top UMOD gene variants, which have also been associated with hypertension and kidney stones, thus highlighting the diverse and significant impact of uromodulin on kidney-related conditions. UMOD gene mutations are implicated in uromodulin-associated kidney disease, while polymorphisms in the UMOD gene show a significant association with CKD. In conclusion, uromodulin holds great promise as an informative biomarker, providing valuable insights into kidney function and disease progression in various clinical scenarios. The identification of UMOD gene variants further strengthens its relevance as a potential target for better understanding kidney-related pathologies and devising novel therapeutic strategies. Future investigations into the roles of uromodulin and regulatory mechanisms are likely to yield even more profound implications for kidney disease diagnosis, risk assessment, and management.
Collapse
Affiliation(s)
- Raïsa Thielemans
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
| | | | - Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
| | - Sander De Bruyne
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.D.B.); (M.O.)
| | - Matthijs Oyaert
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.D.B.); (M.O.)
| | - Marijn M. Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
- Research Foundation Flanders, 1000 Brussels, Belgium
| |
Collapse
|
7
|
Peres RAS, Peruchetti DB, Silva-Aguiar RP, Teixeira DE, Gomes CP, Takiya CM, Pinheiro AAS, Caruso-Neves C. Rapamycin treatment induces tubular proteinuria: role of megalin-mediated protein reabsorption. Front Pharmacol 2023; 14:1194816. [PMID: 37484026 PMCID: PMC10359992 DOI: 10.3389/fphar.2023.1194816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction: Rapamycin is an immunosuppressor that acts by inhibiting the serine/threonine kinase mechanistic target of rapamycin complex 1. Therapeutic use of rapamycin is limited by its adverse effects. Proteinuria is an important marker of kidney damage and a risk factor for kidney diseases progression and has been reported in patients and animal models treated with rapamycin. However, the mechanism underlying proteinuria induced by rapamycin is still an open matter. In this work, we investigated the effects of rapamycin on parameters of renal function and structure and on protein handling by proximal tubule epithelial cells (PTECs). Methods: Healthy BALB/c mice were treated with 1.5 mg/kg rapamycin by oral gavage for 1, 3, or 7 days. At the end of each treatment, the animals were kept in metabolic cages and renal function and structural parameters were analyzed. LLC-PK1 cell line was used as a model of PTECs to test specific effect of rapamycin. Results: Rapamycin treatment did not change parameters of glomerular structure and function. Conversely, there was a transient increase in 24-h proteinuria, urinary protein to creatinine ratio (UPCr), and albuminuria in the groups treated with rapamycin. In accordance with these findings, rapamycin treatment decreased albumin-fluorescein isothiocyanate uptake in the renal cortex. This effect was associated with reduced brush border expression and impaired subcellular distribution of megalin in PTECs. The effect of rapamycin seems to be specific for albumin endocytosis machinery because it did not modify renal sodium handling or (Na++K+)ATPase activity in BALB/c mice and in the LLC-PK1 cell line. A positive Pearson correlation was found between megalin expression and albumin uptake while an inverse correlation was shown between albumin uptake and UPCr or 24-h proteinuria. Despite its effect on albumin handling in PTECs, rapamycin treatment did not induce tubular injury measured by interstitial space and collagen deposition. Conclusion: These findings suggest that proteinuria induced by rapamycin could have a tubular rather than a glomerular origin. This effect involves a specific change in protein endocytosis machinery. Our results open new perspectives on understanding the undesired effect of proteinuria generated by rapamycin.
Collapse
Affiliation(s)
- Rodrigo A. S. Peres
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B. Peruchetti
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo P. Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E. Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos P. Gomes
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- School of Medicine and Surgery, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina M. Takiya
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Acacia S. Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Al-Bataineh MM, Kinlough CL, Marciszyn A, Lam T, Ye L, Kidd K, Maggiore JC, Poland PA, Kmoch S, Bleyer A, Bain DJ, Montalbetti N, Kleyman TR, Hughey RP, Ray EC. Influence of glycoprotein MUC1 on trafficking of the Ca 2+-selective ion channels, TRPV5 and TRPV6, and on in vivo calcium homeostasis. J Biol Chem 2023; 299:102925. [PMID: 36682497 PMCID: PMC9996365 DOI: 10.1016/j.jbc.2023.102925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Polymorphism of the gene encoding mucin 1 (MUC1) is associated with skeletal and dental phenotypes in human genomic studies. Animals lacking MUC1 exhibit mild reduction in bone density. These phenotypes could be a consequence of modulation of bodily Ca homeostasis by MUC1, as suggested by the previous observation that MUC1 enhances cell surface expression of the Ca2+-selective channel, TRPV5, in cultured unpolarized cells. Using biotinylation of cell surface proteins, we asked whether MUC1 influences endocytosis of TRPV5 and another Ca2+-selective TRP channel, TRPV6, in cultured polarized epithelial cells. Our results indicate that MUC1 reduces endocytosis of both channels, enhancing cell surface expression. Further, we found that mice lacking MUC1 lose apical localization of TRPV5 and TRPV6 in the renal tubular and duodenal epithelium. Females, but not males, lacking MUC1 exhibit reduced blood Ca2+. However, mice lacking MUC1 exhibited no differences in basal urinary Ca excretion or Ca retention in response to PTH receptor signaling, suggesting compensation by transport mechanisms independent of TRPV5 and TRPV6. Finally, humans with autosomal dominant tubulointerstitial kidney disease due to frame-shift mutation of MUC1 (ADTKD-MUC1) exhibit reduced plasma Ca concentrations compared to control individuals with mutations in the gene encoding uromodulin (ADTKD-UMOD), consistent with MUC1 haploinsufficiency causing reduced bodily Ca2+. In summary, our results provide further insight into the role of MUC1 in Ca2+-selective TRP channel endocytosis and the overall effects on Ca concentrations.
Collapse
Affiliation(s)
- Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carol L Kinlough
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Allison Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tracey Lam
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lorena Ye
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kendrah Kidd
- Section on Nephrology, Department of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Joseph C Maggiore
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul A Poland
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stanislav Kmoch
- Section on Nephrology, Department of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anthony Bleyer
- Section on Nephrology, Department of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Daniel J Bain
- Department of Geology and Environmental Science, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rebecca P Hughey
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Evan C Ray
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Luda KM, Da Silva C, Ahmadi F, Mowat AM, Ohno H, Kotarsky K, Agace WW. Identification and characterization of murine glycoprotein 2-expressing intestinal dendritic cells. Scand J Immunol 2022; 96:e13219. [PMID: 37807915 PMCID: PMC9786990 DOI: 10.1111/sji.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
The intestinal lamina propria (LP) contains distinct subsets of classical dendritic cells (cDC), each playing key non-redundant roles in intestinal immune homeostasis. Here, we show that glycoprotein 2 (GP2), a GPI-anchored protein and receptor for bacterial type-I fimbriae, is selectively expressed by CD103+CD11b+ cDC in the murine small intestine (SI). GP2 expression was induced on CD103+CD11b+ cDC within the SI-LP and was regulated by IRF4, TGFβR1- and retinoic acid signalling. Mice selectively lacking Gp2 on CD103+CD11b+ cDC (huLang-Cre.gp2fl/fl mice) had normal numbers and proportions of innate and adaptive immune cells in the SI-LP suggesting that GP2 expression by CD103+CD11b+ cDC is not required for intestinal immune homoeostasis.
Collapse
Affiliation(s)
- Katarzyna M Luda
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Clement Da Silva
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fatemeh Ahmadi
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Allan Mcl Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Knut Kotarsky
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - William W Agace
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Mucosal Immunology Laboratory, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
10
|
Zannad F, Ferreira JP, Butler J, Filippatos G, Januzzi JL, Sumin M, Zwick M, Saadati M, Pocock SJ, Sattar N, Anker SD, Packer M. Effect of empagliflozin on circulating proteomics in heart failure: mechanistic insights into the EMPEROR programme. Eur Heart J 2022; 43:4991-5002. [PMID: 36017745 PMCID: PMC9769969 DOI: 10.1093/eurheartj/ehac495] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/15/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
AIMS Sodium-glucose co-transporter 2 (SGLT2) inhibitors improve cardiovascular outcomes in diverse patient populations, but their mechanism of action requires further study. The aim is to explore the effect of empagliflozin on the circulating levels of intracellular proteins in patients with heart failure, using large-scale proteomics. METHODS AND RESULTS Over 1250 circulating proteins were measured at baseline, Week 12, and Week 52 in 1134 patients from EMPEROR-Reduced and EMPEROR-Preserved, using the Olink® Explore 1536 platform. Statistical and bioinformatical analyses identified differentially expressed proteins (empagliflozin vs. placebo), which were then linked to demonstrated biological actions in the heart and kidneys. At Week 12, 32 of 1283 proteins fulfilled our threshold for being differentially expressed, i.e. their levels were changed by ≥10% with a false discovery rate <1% (empagliflozin vs. placebo). Among these, nine proteins demonstrated the largest treatment effect of empagliflozin: insulin-like growth factor-binding protein 1, transferrin receptor protein 1, carbonic anhydrase 2, erythropoietin, protein-glutamine gamma-glutamyltransferase 2, thymosin beta-10, U-type mitochondrial creatine kinase, insulin-like growth factor-binding protein 4, and adipocyte fatty acid-binding protein 4. The changes of the proteins from baseline to Week 52 were generally concordant with the changes from the baseline to Week 12, except empagliflozin reduced levels of kidney injury molecule-1 by ≥10% at Week 52, but not at Week 12. The most common biological action of differentially expressed proteins appeared to be the promotion of autophagic flux in the heart, kidney or endothelium, a feature of 6 proteins. Other effects of differentially expressed proteins on the heart included the reduction of oxidative stress, inhibition of inflammation and fibrosis, and the enhancement of mitochondrial health and energy, repair, and regenerative capacity. The actions of differentially expressed proteins in the kidney involved promotion of autophagy, integrity and regeneration, suppression of renal inflammation and fibrosis, and modulation of renal tubular sodium reabsorption. CONCLUSIONS Changes in circulating protein levels in patients with heart failure are consistent with the findings of experimental studies that have shown that the effects of SGLT2 inhibitors are likely related to actions on the heart and kidney to promote autophagic flux, nutrient deprivation signalling and transmembrane sodium transport.
Collapse
Affiliation(s)
- Faiez Zannad
- Corresponding author. Tel: +33 3 83 15 73 15, Fax: +33 3 83 15 73 24, Emails: ;
| | - João Pedro Ferreira
- Corresponding author. Tel: +33 3 83 15 73 15, Fax: +33 3 83 15 73 24, Emails: ;
| | - Javed Butler
- Heart and Vascular Research, Baylor Scott and White Research Institute, 34 Live Oak St Ste 501, Dallas, TX 75204, USA,University of Mississippi Medical Center, 2500 North State Street Jackson, MS 39216, USA
| | - Gerasimos Filippatos
- Heart Failure Unit, National and Kapodistrian University of Athens School of Medicine, Mikras Asias 75, Athina 115 27 Athens, Greece
| | - James L Januzzi
- Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114USA,The Baim Institute for Clinical Research, 930 Commonwealth Ave #3, Boston, MA 02215USA
| | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Binger Str. 173, 55218 Ingelheim am RheinGermany
| | - Matthias Zwick
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der RissGermany
| | - Maral Saadati
- Elderbrook Solutions GmbH on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riss, Germany
| | - Stuart J Pocock
- London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HTUK
| | - Naveed Sattar
- BHF, UK School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TAUK
| | - Stefan D Anker
- Department of Cardiology (CVK) Berlin Institute of Health Center for Regenerative Therapies (BCRT) German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany,Institute of Heart Diseases, Wroclaw Medical University, Borowska Street 213, 50-556 Warsaw, Poland
| | - Milton Packer
- Baylor Heart and Vascular Hospital, Baylor University Medical Center, 621 N Hall St, Dallas, TX 75226, USA,Imperial College, London, Exhibition Rd, South Kensington, London SW7 2BX, UK
| |
Collapse
|
11
|
Whitehead M, Shanahan CM. Circulating uromodulin: a cytokine trap for osteoinductive inflammatory mediators in chronic kidney disease? Cardiovasc Res 2021; 117:651-652. [PMID: 33367494 DOI: 10.1093/cvr/cvaa348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Meredith Whitehead
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Catherine M Shanahan
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
12
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
13
|
Houser MC, Mac V, Smith DJ, Chicas RC, Xiuhtecutli N, Flocks JD, Elon L, Tansey MG, Sands JM, McCauley L, Hertzberg VS. Inflammation-Related Factors Identified as Biomarkers of Dehydration and Subsequent Acute Kidney Injury in Agricultural Workers. Biol Res Nurs 2021; 23:676-688. [PMID: 34018403 DOI: 10.1177/10998004211016070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Globally, there is increasing recognition that agricultural workers are at risk for chronic kidney disease of unknown etiology (CKDu). Recurrent heat exposure, physical exertion, dehydration, muscle damage, and inflammation are hypothesized to contribute to the development of CKDu, but the relative importance of these processes and the interactions among them remain unclear. Moreover, there is a need to identify biomarkers that could distinguish individuals who are at greatest risk for kidney damage to target preventative interventions for CKDu. In this study, we evaluated dehydration and markers of inflammation, muscle damage, and renal function in agricultural workers at a non-workday baseline assessment. Urine specific gravity and kidney function were measured before and after work shifts on three subsequent days, and heat index, core body temperature, and heart rate were monitored during the work shifts. A combination of direct comparisons and machine learning algorithms revealed that reduced levels of uromodulin and sodium in urine and increased levels of interleukin-6 and C-reactive protein in serum were indicative of dehydration at baseline, and that dehydration, high body mass index, reduced urine uromodulin, and increased serum interleukin-6, C-reactive protein, and lipopolysaccharide-binding protein at baseline were predictive of acute kidney injury on subsequent workdays. Our findings suggest a method for identifying agricultural workers at greatest risk for kidney injury and reveal potential mechanisms responsible for this process, including pathways overlapping in dehydration and kidney injury. These results will guide future studies confirming these mechanisms and introducing interventions to protect kidney health in this vulnerable population.
Collapse
Affiliation(s)
- Madelyn C Houser
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Valerie Mac
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Daniel J Smith
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Roxana C Chicas
- Renal Division, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Nezahualcoyotl Xiuhtecutli
- Farmworker Association of Florida, Apopka, FL, USA.,Department of Anthropology, Tulane University, New Orleans, LA, USA
| | - Joan D Flocks
- Social Policy Division, Center for Governmental Responsibility, Levin College of Law, University of Florida, Gainesville, FL, USA
| | - Lisa Elon
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | | | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Linda McCauley
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Vicki S Hertzberg
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| |
Collapse
|
14
|
Alesutan I, Luong TTD, Schelski N, Masyout J, Hille S, Schneider MP, Graham D, Zickler D, Verheyen N, Estepa M, Pasch A, Maerz W, Tomaschitz A, Pilz S, Frey N, Lang F, Delles C, Müller OJ, Pieske B, Eckardt KU, Scherberich J, Voelkl J. Circulating uromodulin inhibits vascular calcification by interfering with pro-inflammatory cytokine signalling. Cardiovasc Res 2021; 117:930-941. [PMID: 32243494 DOI: 10.1093/cvr/cvaa081] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/15/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Uromodulin is produced exclusively in the kidney and secreted into both urine and blood. Serum levels of uromodulin are correlated with kidney function and reduced in chronic kidney disease (CKD) patients, but physiological functions of serum uromodulin are still elusive. This study investigated the role of uromodulin in medial vascular calcification, a key factor associated with cardiovascular events and mortality in CKD patients. METHODS AND RESULTS Experiments were performed in primary human (HAoSMCs) and mouse (MOVAS) aortic smooth muscle cells, cholecalciferol overload and subtotal nephrectomy mouse models and serum from CKD patients. In three independent cohorts of CKD patients, serum uromodulin concentrations were inversely correlated with serum calcification propensity. Uromodulin supplementation reduced phosphate-induced osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs. In human serum, pro-inflammatory cytokines tumour necrosis factor α (TNFα) and interleukin-1β (IL-1β) co-immunoprecipitated with uromodulin. Uromodulin inhibited TNFα and IL-1β-induced osteo-/chondrogenic signalling and activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated β cells (NF-kB) as well as phosphate-induced NF-kB-dependent transcriptional activity in HAoSMCs. In vivo, adeno-associated virus (AAV)-mediated overexpression of uromodulin ameliorated vascular calcification in mice with cholecalciferol overload. Conversely, cholecalciferol overload-induced vascular calcification was aggravated in uromodulin-deficient mice. In contrast, uromodulin overexpression failed to reduce vascular calcification during renal failure in mice. Carbamylated uromodulin was detected in serum of CKD patients and uromodulin carbamylation inhibited its anti-calcific properties in vitro. CONCLUSIONS Uromodulin counteracts vascular osteo-/chondrogenic transdifferentiation and calcification, at least in part, through interference with cytokine-dependent pro-calcific signalling. In CKD, reduction and carbamylation of uromodulin may contribute to vascular pathology.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Aorta/immunology
- Aorta/metabolism
- Cell Transdifferentiation/drug effects
- Cells, Cultured
- Chondrogenesis
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Humans
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Osteogenesis
- Phenotype
- Protein Carbamylation
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/immunology
- Signal Transduction
- Uromodulin/blood
- Uromodulin/genetics
- Uromodulin/pharmacology
- Vascular Calcification/blood
- Vascular Calcification/immunology
- Vascular Calcification/prevention & control
- Young Adult
- Mice
Collapse
Affiliation(s)
- Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch 2, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Trang T D Luong
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nadeshda Schelski
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jaber Masyout
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus P Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
- German Chronic Kidney Disease (GCKD) Study
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nicolas Verheyen
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Calciscon AG, Aarbergstrasse 5, 2560 Nidau-Biel, Switzerland
- Nierenpraxis Bern, Bubenbergplatz 5, 3011 Bern, Switzerland
- Department of Nephrology, Lindenhofspital, Bremgartenstrasse 117, 3001 Bern, Switzerland
| | - Winfried Maerz
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Ludolf Krehl Street 7-11, 68167 Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, P5,7, 68161 Mannheim, Germany
| | | | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Florian Lang
- Department of Physiology, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch 2, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
- German Chronic Kidney Disease (GCKD) Study
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juergen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München-Harlaching, Teaching Hospital of the Ludwig-Maximilians-Universität, Sanatoriumsplatz 2, 81545 München, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
15
|
Abstract
Uromodulin, a protein exclusively produced by the kidney, is the most abundant urinary protein in physiological conditions. Already described several decades ago, uromodulin has gained the spotlight in recent years, since the discovery that mutations in its encoding gene UMOD cause a renal Mendelian disease (autosomal dominant tubulointerstitial kidney disease) and that common polymorphisms are associated with multifactorial disorders, such as chronic kidney disease, hypertension, and cardiovascular diseases. Moreover, variations in uromodulin levels in urine and/or blood reflect kidney functioning mass and are of prognostic value for renal function, cardiovascular events, and overall mortality. The clinical relevance of uromodulin reflects its multifunctional nature, playing a role in renal ion transport and immunomodulation, in protection against urinary tract infections and renal stones, and possibly as a systemic antioxidant. Here, we discuss the multifaceted roles of this protein in kidney physiology and its translational relevance.
Collapse
Affiliation(s)
- Céline Schaeffer
- Molecular Genetics of Renal Disorders, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy;
| | - Olivier Devuyst
- Mechanisms of Inherited Kidney Disorders Group, University of Zurich, CH-8057 Zurich, Switzerland
| | - Luca Rampoldi
- Molecular Genetics of Renal Disorders, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy;
| |
Collapse
|
16
|
Steubl D, Buzkova P, Garimella PS, Ix JH, Devarajan P, Bennett MR, Chaves PHM, Shlipak MG, Bansal N, Sarnak MJ. Association of serum uromodulin with mortality and cardiovascular disease in the elderly-the Cardiovascular Health Study. Nephrol Dial Transplant 2020; 35:1399-1405. [PMID: 30903163 DOI: 10.1093/ndt/gfz008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/09/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Uromodulin (UMOD) is released by renal tubular cells into the serum (sUMOD) and urine. Lower urine UMOD has been linked to mortality and cardiovascular disease but much less is known about sUMOD. We evaluated the association of sUMOD with these outcomes in community-dwelling older adults. METHODS We measured sUMOD in a random subcohort of 933 participants enrolled in the Cardiovascular Health Study. The associations of sUMOD with all-cause mortality, incident heart failure (HF) and incident cardiovascular disease (CVD; myocardial infarction, stroke and mortality due to coronary disease or stroke) were evaluated using multivariable Cox regression, adjusting for study participants' demographics, estimated glomerular filtration rate (eGFR), albuminuria and CVD risk factors. Generalized additive models with splines were used to address the functional form of sUMOD with outcomes. Due to nonlinear associations of sUMOD with all outcomes, 2.5% of the values on either end of the sUMOD distribution were excluded from the analyses, limiting the range of sUMOD to 34.3-267.1 ng/mL. RESULTS The mean age was 78 ± 5 years, 40% were male, sUMOD level was 127 ± 64 ng/mL, eGFR was 63 mL/min/1.73 m2 and 42% had CKD defined as eGFR <60 mL/min/1.73 m2. Patients in the lower sUMOD quartiles had lower eGFR and higher albuminuria (P < 0.01, respectively). During a median follow-up of 9.9 years, 805 patients died, 283 developed HF and 274 developed CVD. In multivariable analysis, higher sUMOD was significantly associated with a lower hazard for mortality {hazard ratio [HR] 0.89 [95% confidence interval (CI) 0.80-0.99] per 1 standard deviation (SD) higher sUMOD}, CVD [HR 0.80 (95% CI 0.67-0.96)] and the composite endpoint [HR 0.88 (95% CI 0.78-0.99)]; the association with HF was not statistically significant [HR 0.84 (95% CI 0.70-1.01)]. CONCLUSION Higher sUMOD is independently associated with a lower risk for mortality and CVD in older adults.
Collapse
Affiliation(s)
- Dominik Steubl
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Tufts University, Boston, MA, USA.,Division of Nephrology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Petra Buzkova
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Pranav S Garimella
- Division of Nephrology, University of California, San Diego, San Diego, CA, USA
| | - Joachim H Ix
- Division of Nephrology, University of California, San Diego, San Diego, CA, USA
| | - Prasad Devarajan
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Michael R Bennett
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Paulo H M Chaves
- Benjamin Leon Center for Geriatric Research and Education, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Michael G Shlipak
- Division of General Internal Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nisha Bansal
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, WA, USA
| | - Mark J Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Tufts University, Boston, MA, USA
| |
Collapse
|
17
|
LaFavers KA, Macedo E, Garimella PS, Lima C, Khan S, Myslinski J, McClintick J, Witzmann FA, Winfree S, Phillips CL, Hato T, Dagher PC, Wu XR, El-Achkar TM, Micanovic R. Circulating uromodulin inhibits systemic oxidative stress by inactivating the TRPM2 channel. Sci Transl Med 2020; 11:11/512/eaaw3639. [PMID: 31578243 DOI: 10.1126/scitranslmed.aaw3639] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/26/2019] [Accepted: 08/22/2019] [Indexed: 12/21/2022]
Abstract
High serum concentrations of kidney-derived protein uromodulin [Tamm-Horsfall protein (THP)] have recently been shown to be independently associated with low mortality in both older adults and cardiac patients, but the underlying mechanism remains unclear. Here, we show that THP inhibits the generation of reactive oxygen species (ROS) both in the kidney and systemically. Consistent with this experimental data, the concentration of circulating THP in patients with surgery-induced acute kidney injury (AKI) correlated with systemic oxidative damage. THP in the serum dropped after AKI and was associated with an increase in systemic ROS. The increase in oxidant injury correlated with postsurgical mortality and need for dialysis. Mechanistically, THP inhibited the activation of the transient receptor potential cation channel, subfamily M, member 2 (TRPM2) channel. Furthermore, inhibition of TRPM2 in vivo in a mouse model mitigated the systemic increase in ROS during AKI and THP deficiency. Our results suggest that THP is a key regulator of systemic oxidative stress by suppressing TRPM2 activity, and our findings might help explain how circulating THP deficiency is linked with poor outcomes and increased mortality.
Collapse
Affiliation(s)
- Kaice A LaFavers
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Etienne Macedo
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Pranav S Garimella
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Camila Lima
- Division of Nephrology, Department of Medicine, University of Sao Paulo, Sao Paulo 05403, Brazil
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jered Myslinski
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeanette McClintick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Frank A Witzmann
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Seth Winfree
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carrie L Phillips
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Takashi Hato
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pierre C Dagher
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine and Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA. .,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Radmila Micanovic
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
Then C, Herder C, Then H, Thorand B, Huth C, Heier M, Meisinger C, Peters A, Koenig W, Rathmann W, Roden M, Stumvoll M, Maalmi H, Meitinger T, Lechner A, Scherberich J, Seissler J. Serum uromodulin is inversely associated with biomarkers of subclinical inflammation in the population-based KORA F4 study. Clin Kidney J 2020; 14:1618-1625. [PMID: 34221377 PMCID: PMC8248959 DOI: 10.1093/ckj/sfaa165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Indexed: 01/24/2023] Open
Abstract
Background Uromodulin is a kidney-specific glycoprotein synthesized in tubular cells of
Henle’s loop exerting nephroprotective and immunomodulatory
functions in the urinary tract. A small amount of uromodulin is also
released into the systemic circulation, where its physiological role is
unknown. Serum uromodulin (sUmod) has been associated with metabolic risk
factors and with cardiovascular events and mortality, where these
associations were partly stronger in men than in women. In this study, we
investigated the associations of sUmod with biomarkers of subclinical
inflammation in a population-based sample of women and men. Methods Associations of sUmod with 10 biomarkers of subclinical inflammation were
assessed in 1065 participants of the Cooperative Health Research in the
Region of Augsburg (KORA) F4 study aged 62–81 years using
linear regression models adjusted for sex, age, body mass index, estimated
glomerular filtration rate and diabetes. Analyses were performed in the
total study sample and stratified by sex. Results sUmod was inversely associated with white blood cell count, high-sensitive
C-reactive protein, interleukin (IL)-6, tumour necrosis factor-α,
myeloperoxidase, superoxide dismutase-3, IL-1 receptor antagonist and IL-22
after multivariable adjustment and correction for multiple testing
(P < 0.001 for each observation). There was a trend
towards a stronger association of sUmod with pro-inflammatory markers in men
than in women, with a significant P for sex interaction (<0.001)
regarding the relation of sUmod with IL-6. Conclusions sUmod was inversely associated with biomarkers of subclinical inflammation in
older participants of the KORA F4 study. The association of sUmod with IL-6
differed between women and men. Future research should focus on whether the
immunomodulatory properties of sUmod are one explanation for the association
of sUmod with cardiovascular outcomes and mortality.
Collapse
Affiliation(s)
- Cornelia Then
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU, München, Germany.,Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Holger Then
- Mathematics department, Freie Waldorfschule Augsburg, Augsburg, Germany
| | - Barbara Thorand
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Cornelia Huth
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,KORA Study Centre, University Hospital Augsburg, Augsburg, Germany
| | - Christa Meisinger
- Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,Chair of Epidemiology at UNIKAT Augsburg, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Wolfgang Koenig
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany.,Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,German Diabetes Center, Leibniz Institute at Heinrich Heine University Düsseldorf, Institute of Biometrics and Epidemiology, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | | - Haifa Maalmi
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Meitinger
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Andreas Lechner
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU, München, Germany.,Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Jürgen Scherberich
- Klinikum München-Harlaching, Teaching Hospital of the Ludwig-Maximilians-Universität, Munich, Germany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU, München, Germany.,Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
19
|
Micanovic R, LaFavers K, Garimella PS, Wu XR, El-Achkar TM. Uromodulin (Tamm-Horsfall protein): guardian of urinary and systemic homeostasis. Nephrol Dial Transplant 2020; 35:33-43. [PMID: 30649494 DOI: 10.1093/ndt/gfy394] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022] Open
Abstract
Biology has taught us that a protein as abundantly made and conserved among species as Tamm-Horsfall protein (THP or uromodulin) cannot just be a waste product serving no particular purpose. However, for many researchers, THP is merely a nuisance during urine proteome profiling or exosome purification and for clinicians an enigmatic entity without clear disease implications. Thanks to recent human genetic and correlative studies and animal modeling, we now have a renewed appreciation of this highly prevalent protein in not only guarding urinary homeostasis, but also serving as a critical mediator in systemic inter-organ signaling. Beyond a mere barrier that lines the tubules, or a surrogate for nephron mass, mounting evidence suggests that THP is a multifunctional protein critical for modulating renal ion channel activity, salt/water balance, renal and systemic inflammatory response, intertubular communication, mineral crystallization and bacterial adhesion. Indeed, mutations in THP cause a group of inherited kidney diseases, and altered THP expression is associated with increased risks of urinary tract infection, kidney stone, hypertension, hyperuricemia and acute and chronic kidney diseases. Despite the recent surge of information surrounding THP's physiological functions and disease involvement, our knowledge remains incomplete regarding how THP is normally regulated by external and intrinsic factors, how precisely THP deficiency leads to urinary and systemic pathophysiology and in what clinical settings THP can be used as a theranostic biomarker and a target for modulation to improve patient outcomes.
Collapse
Affiliation(s)
- Radmila Micanovic
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kaice LaFavers
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pranav S Garimella
- Department of Medicine, Division of Nephrology-Hypertension, University of California, San Diego, San Diego, CA, USA
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine, New York, NY, USA.,Veterans Affairs New York Harbor Healthcare System, New York City, NY, USA
| | - Tarek M El-Achkar
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Uromodulin (UMOD), also known as Tamm-Horsfall protein, is the most abundant protein in human urine. UMOD has multiple functions such as protection against urinary tract infections and nephrolithiasis. This review outlines recent progress made in UMOD's role in renal physiology, tubular transport, and mineral metabolism. RECENT FINDINGS UMOD is mostly secreted in the thick ascending limb (TAL) and to a lesser degree in the distal convoluted tubule (DCT). UMOD secretion is regulated by the calcium-sensing receptor. UMOD upregulates ion channels [e.g., renal outer medullary potassium channel, transient receptor potential cation channel subfamily V member 5, and transient receptor potential melastatin 6 (TRPM6)] and cotransporters [e.g., Na,K,2Cl cotransporter (NKCC2) and sodium-chloride cotransporter (NCC)] in the TAL and DCT. Higher serum UMOD concentrations have been associated with higher renal function and preserved renal reserve. Higher serum UMOD has also been linked to a lower risk of cardiovascular disease and diabetes mellitus. SUMMARY With better serum UMOD detection assays the extent of different functions for UMOD is still expanding. Urinary UMOD regulates different tubular ion channels and cotransporters. Variations of urinary UMOD secretion can so contribute to common disorders such as hypertension or nephrolithiasis.
Collapse
|
21
|
Friedman DJ, Alper SL. Modulation of tubular solute reuptake in UMOD knockout mice. Am J Physiol Renal Physiol 2018. [PMID: 29513073 DOI: 10.1152/ajprenal.00080.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- David J Friedman
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School , Boston, Massachusetts
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
22
|
Wu TH, Li KJ, Yu CL, Tsai CY. Tamm-Horsfall Protein is a Potent Immunomodulatory Molecule and a Disease Biomarker in the Urinary System. Molecules 2018; 23:molecules23010200. [PMID: 29361765 PMCID: PMC6017547 DOI: 10.3390/molecules23010200] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/17/2018] [Accepted: 01/17/2018] [Indexed: 01/14/2023] Open
Abstract
Tamm–Horsfall protein (THP), or uromodulin (UMOD), is an 80–90-kDa phosphatidylinositol-anchored glycoprotein produced exclusively by the renal tubular cells in the thick ascending limb of the loop of Henle. Physiologically, THP is implicated in renal countercurrent gradient formation, sodium homeostasis, blood pressure regulation, and a defense molecule against infections in the urinary system. Investigations have also revealed that THP is an effective binding ligand for serum albumin, immunoglobulin G light chains, complement components C1 and C1q, interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor (TNF)-α, and interferon-γ through its carbohydrate side chains for maintaining circulatory and renal immune homeostasis. Thus, THP can be regarded as part of the innate immune system. UMOD mutations play crucial roles in congenital urolithiasis, hereditary hyperuricemia/gout, and medullary cystic kidney diseases. Recent investigations have focused on the immunomodulatory effects of THP on immune cells and on THP as a disease biomarker of acute and chronic kidney diseases. Our studies have suggested that normal urinary THP, through its epidermal growth factor (EGF)-like domains, binds to the surface-expressed EGF-like receptors, cathepsin G, or lactoferrin to enhance polymorphonuclear leukocyte phagocytosis, proinflammatory cytokine production by monocytes/macrophages, and lymphocyte proliferation by activating the Rho family and mitogen-activated protein kinase signaling pathways. Furthermore, our data support both an intact protein core structure and carbohydrate side chains are important for the different protein-binding capacities of THP. Prospectively, parts of the whole THP molecule may be used for anti-TNF-α therapy in inflammatory diseases, autoantibody-depleting therapy in autoimmune disorders, and immune intensification in immunocompromised hosts.
Collapse
Affiliation(s)
- Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 112, Taiwan.
| | - Ko-Jen Li
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan.
| | - Chia-Li Yu
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan.
| | - Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, 201 Shih-Pai Road, Sec 2, Taipei 112, Taiwan.
| |
Collapse
|
23
|
Liu Y, Goldfarb DS, El-Achkar TM, Lieske JC, Wu XR. Tamm-Horsfall protein/uromodulin deficiency elicits tubular compensatory responses leading to hypertension and hyperuricemia. Am J Physiol Renal Physiol 2018; 314:F1062-F1076. [PMID: 29357410 DOI: 10.1152/ajprenal.00233.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Expression of Tamm-Horsfall protein (THP or uromodulin) is highly restricted to the kidney thick ascending limb (TAL) of loop of Henle. Despite the unique location and recent association of THP gene mutations with hereditary uromodulin-associated kidney disease and THP single nucleotide polymorphisms with chronic kidney disease and hypertension, the physiological function(s) of THP and its pathological involvement remain incompletely understood. By studying age-dependent changes of THP knockout (KO) mice, we show here that young KO mice had significant salt and water wasting but were partially responsive to furosemide, due to decreased luminal translocation of Na-K-Cl cotransporter 2 (NKCC2) in the TAL. Aged THP KO mice were, however, markedly oliguric and unresponsive to furosemide, and their NKCC2 was localized primarily in the cytoplasm as evidenced by lipid raft floatation assay, cell fractionation, and confocal and immunoelectron microscopy. These aged KO mice responded to metolazone and acetazolamide, known to target distal and proximal tubules, respectively. They also had marked upregulation of renin in juxtaglomerular apparatus and serum, and they were hypertensive. Finally, the aged THP KO mice had significant upregulation of Na-coupled urate transporters Slc5a8 and Slc22a12 as well as sodium-hydrogen exchanger 3 (NHE3) in the proximal tubule and elevated serum uric acid and allantoin. Collectively, our results suggest that THP deficiency can cause progressive disturbances in renal functions via initially NKCC2 dysfunction and later compensatory responses, resulting in prolonged activation of the renin-angiotensin-aldosterone axis and hyperuricemia.
Collapse
Affiliation(s)
- Yan Liu
- Department of Urology, New York University School of Medicine , New York, New York
| | - David S Goldfarb
- Department of Nephrology, New York University School of Medicine , New York, New York.,Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, New York
| | - Tarek M El-Achkar
- Division of Nephrology, Indiana University School of Medicine and Indianapolis Veterans Affairs , Indianapolis, Indiana
| | - John C Lieske
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Xue-Ru Wu
- Department of Urology, New York University School of Medicine , New York, New York.,Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, New York.,Department of Pathology, New York University School of Medicine , New York, New York
| |
Collapse
|
24
|
Devuyst O, Olinger E, Rampoldi L. Uromodulin: from physiology to rare and complex kidney disorders. Nat Rev Nephrol 2017; 13:525-544. [PMID: 28781372 DOI: 10.1038/nrneph.2017.101] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Uromodulin (also known as Tamm-Horsfall protein) is exclusively produced in the kidney and is the most abundant protein in normal urine. The function of uromodulin remains elusive, but the available data suggest that this protein might regulate salt transport, protect against urinary tract infection and kidney stones, and have roles in kidney injury and innate immunity. Interest in uromodulin was boosted by genetic studies that reported involvement of the UMOD gene, which encodes uromodulin, in a spectrum of rare and common kidney diseases. Rare mutations in UMOD cause autosomal dominant tubulointerstitial kidney disease (ADTKD), which leads to chronic kidney disease (CKD). Moreover, genome-wide association studies have identified common variants in UMOD that are strongly associated with risk of CKD and also with hypertension and kidney stones in the general population. These findings have opened up a new field of kidney research. In this Review we summarize biochemical, physiological, genetic and pathological insights into the roles of uromodulin; the mechanisms by which UMOD mutations cause ADTKD, and the association of common UMOD variants with complex disorders.
Collapse
Affiliation(s)
- Olivier Devuyst
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Eric Olinger
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Luca Rampoldi
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
25
|
Graham LA, Dominiczak AF, Ferreri NR. Role of renal transporters and novel regulatory interactions in the TAL that control blood pressure. Physiol Genomics 2017; 49:261-276. [PMID: 28389525 PMCID: PMC5451551 DOI: 10.1152/physiolgenomics.00017.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/27/2017] [Accepted: 04/05/2017] [Indexed: 12/31/2022] Open
Abstract
Hypertension (HTN), a major public health issue is currently the leading factor in the global burden of disease, where associated complications account for 9.4 million deaths worldwide every year. Excessive dietary salt intake is among the environmental factors that contribute to HTN, known as salt sensitivity. The heterogeneity of salt sensitivity and the multiple mechanisms that link high salt intake to increases in blood pressure are of upmost importance for therapeutic application. A continual increase in the kidney's reabsorption of sodium (Na+) relies on sequential actions at various segments along the nephron. When the distal segments of the nephron fail to regulate Na+, the effects on Na+ homeostasis are unfavorable. We propose that the specific nephron region where increased active uptake occurs as a result of variations in Na+ reabsorption is at the thick ascending limb of the loop of Henle (TAL). The purpose of this review is to urge the consideration of the TAL as contributing to the pathophysiology of salt-sensitive HTN. Further research in this area will enable development of a therapeutic application for targeted treatment.
Collapse
Affiliation(s)
- Lesley A Graham
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
26
|
Proteus mirabilis fimbriae- and urease-dependent clusters assemble in an extracellular niche to initiate bladder stone formation. Proc Natl Acad Sci U S A 2016; 113:4494-9. [PMID: 27044107 DOI: 10.1073/pnas.1601720113] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The catheter-associated uropathogenProteus mirabilisfrequently causes urinary stones, but little has been known about the initial stages of bladder colonization and stone formation. We found thatP. mirabilisrapidly invades the bladder urothelium, but generally fails to establish an intracellular niche. Instead, it forms extracellular clusters in the bladder lumen, which form foci of mineral deposition consistent with development of urinary stones. These clusters elicit a robust neutrophil response, and we present evidence of neutrophil extracellular trap generation during experimental urinary tract infection. We identified two virulence factors required for cluster development: urease, which is required for urolithiasis, and mannose-resistantProteus-like fimbriae. The extracellular cluster formation byP. mirabilisstands in direct contrast to uropathogenicEscherichia coli, which readily formed intracellular bacterial communities but not luminal clusters or urinary stones. We propose that extracellular clusters are a key mechanism ofP. mirabilissurvival and virulence in the bladder.
Collapse
|
27
|
Kisiela DI, Avagyan H, Friend D, Jalan A, Gupta S, Interlandi G, Liu Y, Tchesnokova V, Rodriguez VB, Sumida JP, Strong RK, Wu XR, Thomas WE, Sokurenko EV. Inhibition and Reversal of Microbial Attachment by an Antibody with Parasteric Activity against the FimH Adhesin of Uropathogenic E. coli. PLoS Pathog 2015; 11:e1004857. [PMID: 25974133 PMCID: PMC4431754 DOI: 10.1371/journal.ppat.1004857] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 04/06/2015] [Indexed: 11/18/2022] Open
Abstract
Attachment proteins from the surface of eukaryotic cells, bacteria and viruses are critical receptors in cell adhesion or signaling and are primary targets for the development of vaccines and therapeutic antibodies. It is proposed that the ligand-binding pocket in receptor proteins can shift between inactive and active conformations with weak and strong ligand-binding capability, respectively. Here, using monoclonal antibodies against a vaccine target protein - fimbrial adhesin FimH of uropathogenic Escherichia coli, we demonstrate that unusually strong receptor inhibition can be achieved by antibody that binds within the binding pocket and displaces the ligand in a non-competitive way. The non-competitive antibody binds to a loop that interacts with the ligand in the active conformation of the pocket but is shifted away from ligand in the inactive conformation. We refer to this as a parasteric inhibition, where the inhibitor binds adjacent to the ligand in the binding pocket. We showed that the receptor-blocking mechanism of parasteric antibody differs from that of orthosteric inhibition, where the inhibitor replaces the ligand or allosteric inhibition where the inhibitor binds at a site distant from the ligand, and is very potent in blocking bacterial adhesion, dissolving surface-adherent biofilms and protecting mice from urinary bladder infection. A common approach in the development of selective inhibitors for ligand-receptor interactions is targeting the receptor binding site with the expectation that inhibitors will sterically interfere with ligand binding and thus block receptor function via a competitive (orthosteric) mechanism. However, using monoclonal antibodies specific for the mannose-binding Escherichia coli adhesin, FimH, we demonstrate that the binding site epitopes allow for non-competitive inhibition that is more effective than orthosteric blocking. FimH, similar to other binding proteins, exhibits conformational flexibility of the ligand-binding pocket shifting between open (inactive) and tight (active) conformations, with relatively low- and high- affinity towards mannose. We show that an antibody that binds just one of the mannose-binding pocket loops prevents the shift from the inactive to the active conformation and hence blocks formation of high-affinity ligand-receptor complexes. This antibody type was more effective in inhibition of bacterial adhesion than anti-FimH antibodies competitively blocking mannose binding, and unlike the latter or a soluble ligand, showed the ability to detach an established bacterial biofilm from a ligand-coated surface. As the newly described antibody can bind the FimH pocket simultaneously with ligand, we refer to it as a parasteric (next-to-ligand) inhibitor that exhibits non-competitive inhibition from within the binding-pocket of the receptor.
Collapse
Affiliation(s)
- Dagmara I. Kisiela
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DIK); (EVS)
| | - Hovhannes Avagyan
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Della Friend
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aachal Jalan
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Shivani Gupta
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Gianluca Interlandi
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Yan Liu
- Department of Urology, New York University School of Medicine, New York, New York, United States of America
| | - Veronika Tchesnokova
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Victoria B. Rodriguez
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - John P. Sumida
- Analytical Biopharmacy Core, University of Washington, Seattle, Washington, United States of America
| | - Roland K. Strong
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Xue-Ru Wu
- Department of Urology, New York University School of Medicine, New York, New York, United States of America
| | - Wendy E. Thomas
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Evgeni V. Sokurenko
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DIK); (EVS)
| |
Collapse
|
28
|
El-Achkar TM, Dagher PC. Tubular cross talk in acute kidney injury: a story of sense and sensibility. Am J Physiol Renal Physiol 2015; 308:F1317-23. [PMID: 25877507 DOI: 10.1152/ajprenal.00030.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/10/2015] [Indexed: 01/20/2023] Open
Abstract
The mammalian kidney is an organ composed of numerous functional units or nephrons. Beyond the filtering glomerulus of each nephron, various tubular segments with distinct populations of epithelial cells sequentially span the kidney from cortex to medulla. The highly organized folding of the tubules results in a spatial distribution that allows intimate contact between various tubular subsegments. This unique arrangement can promote a newly recognized type of horizontal epithelial-to-epithelial cross talk. In this review, we discuss the importance of this tubular cross talk in shaping the response of the kidney to acute injury in a sense and sensibility model. We propose that injury-resistant tubules such as S1 proximal segments and thick ascending limbs (TAL) can act as "sensors" and thus modulate the responsiveness or "sensibility" of the S2-S3 proximal segments to injury. We also discuss new findings that highlight the importance of tubular cross talk in regulating homeostasis and inflammation not only in the kidney, but also systemically.
Collapse
Affiliation(s)
- Tarek M El-Achkar
- Indiana University School of Medicine, Indianapolis, Indiana; and Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Pierre C Dagher
- Indiana University School of Medicine, Indianapolis, Indiana; and
| |
Collapse
|
29
|
Micanovic R, Chitteti BR, Dagher PC, Srour EF, Khan S, Hato T, Lyle A, Tong Y, Wu XR, El-Achkar TM. Tamm-Horsfall Protein Regulates Granulopoiesis and Systemic Neutrophil Homeostasis. J Am Soc Nephrol 2015; 26:2172-82. [PMID: 25556169 DOI: 10.1681/asn.2014070664] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
Abstract
Tamm-Horsfall protein (THP) is a glycoprotein uniquely expressed in the kidney. We recently showed an important role for THP in mediating tubular cross-talk in the outer medulla and in suppressing neutrophil infiltration after kidney injury. However, it remains unclear whether THP has a broader role in neutrophil homeostasis. In this study, we show that THP deficiency in mice increases the number of neutrophils, not only in the kidney but also in the circulation and in the liver, through enhanced granulopoiesis in the bone marrow. Using multiplex ELISA, we identified IL-17 as a key granulopoietic cytokine specifically upregulated in the kidneys but not in the liver of THP(-/-) mice. Indeed, neutralization of IL-17 in THP(-/-) mice completely reversed the systemic neutrophilia. Furthermore, IL-23 was also elevated in THP(-/-) kidneys. We performed real-time PCR on laser microdissected tubular segments and FACS-sorted renal immune cells and identified the S3 proximal segments, but not renal macrophages, as a major source of increased IL-23 synthesis. In conclusion, we show that THP deficiency stimulates proximal epithelial activation of the IL-23/IL-17 axis and systemic neutrophilia. Our findings provide evidence that the kidney epithelium in the outer medulla can regulate granulopoiesis. When this novel function is added to its known role in erythropoiesis, the kidney emerges as an important regulator of the hematopoietic system.
Collapse
Affiliation(s)
| | - Brahmananda R Chitteti
- Hematology, Microbiology, and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Edward F Srour
- Hematology, Microbiology, and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | - Yan Tong
- Divisions of Nephrology and Hematology, Microbiology, and Immunology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biostatistics, Indiana University Schools of Medicine and Public Health, Indianapolis, Indiana; Departments of Urology and Pathology, New York University School of Medicine and Veterans Affairs New York Harbor Healthcare System Manhattan Campus, New York, New York; and Roudebush Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine and Veterans Affairs New York Harbor Healthcare System Manhattan Campus, New York, New York; and
| | - Tarek M El-Achkar
- Divisions of Nephrology and Roudebush Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana
| |
Collapse
|
30
|
Musante L, Tataruch DE, Holthofer H. Use and isolation of urinary exosomes as biomarkers for diabetic nephropathy. Front Endocrinol (Lausanne) 2014; 5:149. [PMID: 25309511 PMCID: PMC4176463 DOI: 10.3389/fendo.2014.00149] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/05/2014] [Indexed: 01/08/2023] Open
Abstract
Diabetes represents a major threat to public health and the number of patients is increasing alarmingly in the global scale. Particularly, the diabetic kidney disease (nephropathy, DN) together with its cardiovascular complications cause immense human suffering, highly increased risk of premature deaths, and lead to huge societal costs. DN is first detected when protein appears in urine (microalbuminuria). As in other persisting proteinuric diseases (like vasculitis) it heralds irreversible damage of kidney functions up to non-functional (end-stage) kidney and ultimately calls for kidney replacement therapy (dialysis or kidney transplantation). While remarkable progress has been made in understanding the genetic and molecular factors associating with chronic kidney diseases, breakthroughs are still missing to provide comprehensive understanding of events and mechanisms associated. Non-invasive diagnostic tools for early diagnostics of kidney damage are badly needed. Exosomes - small vesicular structures present in urine are released by all cell types along kidney structures to present with distinct surface assembly. Furthermore, exosomes carry a load of special proteins and nucleic acids. This "cargo" faithfully reflects the physiological state of their respective cells of origin and appears to serve as a new pathway for downstream signaling to target cells. Accordingly, exosome vesicles are emerging as a valuable source for disease stage-specific information and as fingerprints of disease progression. Unfortunately, technical issues of exosome isolation are challenging and, thus, their full potential remains untapped. Here, we review the molecular basis of exosome secretion as well as their use to reveal events along the nephron. In addition to novel molecular information, the new methods provide the needed accurate, personalized, non-invasive, and inexpensive future diagnostics.
Collapse
Affiliation(s)
- Luca Musante
- Centre for BioAnalytical Sciences (CBAS), Dublin City University, Dublin, Ireland
| | - Dorota Ewa Tataruch
- Centre for BioAnalytical Sciences (CBAS), Dublin City University, Dublin, Ireland
| | - Harry Holthofer
- Centre for BioAnalytical Sciences (CBAS), Dublin City University, Dublin, Ireland
| |
Collapse
|
31
|
Interstitial calcinosis in renal papillae of genetically engineered mouse models: relation to Randall's plaques. Urolithiasis 2014; 43 Suppl 1:65-76. [PMID: 25096800 DOI: 10.1007/s00240-014-0699-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 07/22/2014] [Indexed: 02/08/2023]
Abstract
Genetically engineered mouse models (GEMMs) have been highly instrumental in elucidating gene functions and molecular pathogenesis of human diseases, although their use in studying kidney stone formation or nephrolithiasis remains relatively limited. This review intends to provide an overview of several knockout mouse models that develop interstitial calcinosis in the renal papillae. Included herein are mice deficient for Tamm-Horsfall protein (THP; also named uromodulin), osteopontin (OPN), both THP and OPN, Na(+)-phosphate cotransporter Type II (Npt2a) and Na(+)/H(+) exchanger regulatory factor (NHERF-1). The baseline information of each protein is summarized, along with key morphological features of the interstitial calcium deposits in mice lacking these proteins. Attempts are made to correlate the papillary interstitial deposits found in GEMMs with Randall's plaques, the latter considered precursors of idiopathic calcium stones in patients. The pathophysiology that underlies the renal calcinosis in the knockout mice is also discussed wherever information is available. Not all the knockout models are allocated equal space because some are more extensively characterized than others. Despite the inroads already made, the exact physiological underpinning, origin, evolution and fate of the papillary interstitial calcinosis in the GEMMs remain incompletely defined. Greater investigative efforts are warranted to pin down the precise role of the papillary interstitial calcinosis in nephrolithiasis using the existing models. Additionally, more sophisticated, second-generation GEMMs that allow gene inactivation in a time-controlled manner and "compound mice" that bear several genetic alterations are urgently needed, in light of mounting evidence that nephrolithiasis is a multifactorial, multi-stage and polygenic disease.
Collapse
|
32
|
Uromodulin: old friend with new roles in health and disease. Pediatr Nephrol 2014; 29:1151-8. [PMID: 23880785 DOI: 10.1007/s00467-013-2563-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/16/2013] [Accepted: 06/26/2013] [Indexed: 12/11/2022]
Abstract
The most abundant urinary protein, Tamm-Horsfall protein, later renamed uromodulin, is expressed exclusively by the thick ascending limb cells of the kidney and released into urine from the apical cell membrane. Uromodulin is believed to protect against urinary tract infections and stones, but its other physiologic functions have remained obscure until recently. Renewed interest in uromodulin has been brought about by the identification of uromodulin mutations as causes of a discrete group of diseases that are distinct from nephronophthisis. The three overlapping clinical uromodulin-associated kidney diseases (UAKD) are medullary cystic disease type 2, familial juvenile hyperuricemic nephropathy and glomerulocystic kidney disease. Previously thought of as "adult diseases", it is now recognized that they may also present in childhood and even in infancy. Common characteristics of all three diseases are autosomal dominant inheritance, unremarkable urine sediment and slow progression to end-stage renal disease (ESRD). They are frequently associated with hyperuricemia and gout. These diseases appear to result from failure of the mutant uromodulin to be incorporated into the apical cilium, thereby placing UAKD in the category of "ciliopathies". In addition to causing specific UAKD, certain uromodulin gene polymorphisms have been linked to ESRD in general, suggesting that uromodulin plays a modulatory role in kidney disease progression.
Collapse
|
33
|
Nair AR, Masson GS, Ebenezer PJ, Del Piero F, Francis J. Role of TLR4 in lipopolysaccharide-induced acute kidney injury: protection by blueberry. Free Radic Biol Med 2014; 71:16-25. [PMID: 24657730 DOI: 10.1016/j.freeradbiomed.2014.03.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/19/2014] [Accepted: 03/08/2014] [Indexed: 12/20/2022]
Abstract
Inflammation has been implicated in the pathophysiology of kidney disorders. Previous studies have documented the contributions of various inflammatory cascades in the development of kidney and other organ dysfunctions. The Toll-like receptor 4 (TLR4) inflammatory pathway is a major contributor of inflammation in the kidney. Interestingly, lipopolysaccharide (LPS), a specific ligand for TLR4, has been shown to induce acute kidney injury (AKI) in animal models. We have previously studied the beneficial effects of nonpharmacological agents, particularly blueberries (BB), in attenuating inflammation and oxidative stress. We hypothesize that BB protect against the LPS-induced AKI by inhibiting TLR4 activation and kidney injury markers. Twelve-week-old male Sprague-Dawley rats received a BB solution or saline intragastric gavage for 2 days. One group of BB and saline-gavaged animals was injected with LPS (10 mg/kg bw). Another group of rats was injected with VIPER (0.1 mg/kg iv), a TLR4-specific inhibitory peptide, 2 h before LPS administration. Compared to LPS-administered rats, the BB-pretreated animals exhibited improved glomerular filtration rate, elevated renal blood flow, and a reduced renal vascular resistance. In addition, a reduction in the rate of production of free radicals, namely total reactive oxygen species (ROS) and superoxide, was observed in the BB-supplemented LPS group. Gene and protein expressions for TLR4, proinflammatory cytokine, and acute kidney injury markers were also attenuated in animals that were pretreated with BB as measured by real time RT-PCR and Western blotting, respectively. These results in the BB-pretreated group were consistent with those in the VIPER-treated rats, and indicate that BB protects against AKI by inhibiting TLR4 and its subsequent effect on inflammatory and oxidative stress pathways.
Collapse
Affiliation(s)
- Anand R Nair
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Gustavo S Masson
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Philip J Ebenezer
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fabio Del Piero
- Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Joseph Francis
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
34
|
Mao S, Zhang A, Huang S. The signaling pathway of uromodulin and its role in kidney diseases. J Recept Signal Transduct Res 2014; 34:440-4. [PMID: 24849497 DOI: 10.3109/10799893.2014.920029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The uromodulin (UMOD) is a glycoprotein expressed exclusively by renal tubular cells lining the thick ascending limb of the loop of Henle. UMOD acts as a regulatory protein in health and in various conditions. For kidney diseases, its role remains elusive. On one hand, UMOD plays a role in binding and excretion of various potentially injurious products from the tubular fluid. On the other hand, chronic kidney disease is associated with higher serum levels of UMOD. Signaling pathways might be very important in the pathogenesis of kidney diseases. We performed this review to provide a relatively complete signaling pathway flowchart for UMOD to the investigators who were interested in the role of UMOD in the pathogenesis of kidney diseases. Here, we reviewed the signal transduction pathway of UMOD and its role in the pathogenesis of kidney diseases.
Collapse
Affiliation(s)
- Song Mao
- Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University , Nanjing , China
| | | | | |
Collapse
|
35
|
Kemter E, Prückl P, Rathkolb B, Micklich K, Adler T, Becker L, Beckers J, Busch DH, Götz AA, Hans W, Horsch M, Ivandic B, Klingenspor M, Klopstock T, Rozman J, Schrewe A, Schulz H, Fuchs H, Gailus-Durner V, Hrabé de Angelis M, Wolf E, Aigner B. Standardized, systemic phenotypic analysis of Umod(C93F) and Umod(A227T) mutant mice. PLoS One 2013; 8:e78337. [PMID: 24205203 PMCID: PMC3813435 DOI: 10.1371/journal.pone.0078337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/18/2013] [Indexed: 12/31/2022] Open
Abstract
Uromodulin-associated kidney disease (UAKD) summarizes different clinical features of an autosomal dominant heritable disease syndrome in humans with a proven uromodulin (UMOD) mutation involved. It is often characterized by hyperuricemia, gout, alteration of urine concentrating ability, as well as a variable rate of disease progression inconstantly leading to renal failure and histological alterations of the kidneys. We recently established the two Umod mutant mouse lines Umod(C93F) and Umod(A227T) on the C3H inbred genetic background both showing kidney defects analogous to those found in human UAKD patients. In addition, disease symptoms were revealed that were not yet described in other published mouse models of UAKD. To examine if further organ systems and/or metabolic pathways are affected by Umod mutations as primary or secondary effects, we describe a standardized, systemic phenotypic analysis of the two mutant mouse lines Umod(A227T) and Umod(C93F) in the German Mouse Clinic. Different genotypes as well as different ages were tested. Beside the already published changes in body weight, body composition and bone metabolism, the influence of the Umod mutation on energy metabolism was confirmed. Hematological analysis revealed a moderate microcytic and erythropenic anemia in older Umod mutant mice. Data of the other analyses in 7-10 month-old mutant mice showed single small additional effects.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Petra Prückl
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kateryna Micklich
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thure Adler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Microbiology, Immunology, and Hygiene, TU, Munich, Munich, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Neurology, Friedrich-Baur-Institut, LMU, Munich, Munich, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, TU, Munich, Freising-Weihenstephan, Germany
| | - Dirk H. Busch
- Institute of Medical Microbiology, Immunology, and Hygiene, TU, Munich, Munich, Germany
| | - Alexander A. Götz
- German Mouse Clinic, Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marion Horsch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Boris Ivandic
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, TU, Munich, Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institut, LMU, Munich, Munich, Germany
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Campus Grosshadern, Munich, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, TU, Munich, Freising-Weihenstephan, Germany
| | - Anja Schrewe
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Hrabé de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, TU, Munich, Freising-Weihenstephan, Germany
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Campus Grosshadern, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Bernhard Aigner
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
36
|
Kemter E, Prueckl P, Sklenak S, Rathkolb B, Habermann FA, Hans W, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Wolf E, Aigner B, Wanke R. Type of uromodulin mutation and allelic status influence onset and severity of uromodulin-associated kidney disease in mice. Hum Mol Genet 2013; 22:4148-63. [PMID: 23748428 DOI: 10.1093/hmg/ddt263] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Uromodulin-associated kidney disease (UAKD) is a dominant heritable renal disease in humans which is caused by mutations in the uromodulin (UMOD) gene and characterized by heterogeneous clinical appearance. To get insights into possible causes of this heterogeneity of UAKD, we describe the new mutant mouse line Umod(C93F), leading to disruption of a putative disulfide bond which is also absent in a known human UMOD mutation, and compare the phenotype of this new mouse line with the recently published mouse line Umod(A227T). In both mutant mouse lines, which were both bred on the C3H background, the Umod mutations cause a gain-of-toxic function due to a maturation defect of the mutant uromodulin leading to a dysfunction of thick ascending limb of Henle's loop (TALH) cells of the kidney. Umod mutant mice exhibit increased plasma urea and Cystatin levels, impaired urinary concentration ability, reduced fractional excretion of uric acid and nephropathological alterations including uromodulin retention in TALH cells, interstitial fibrosis and inflammatory cell infiltrations, tubular atrophy and occasional glomerulo- und tubulocystic changes, a phenotype highly similar to UAKD in humans. The maturation defect of mutant uromodulin leads to the accumulation of immature uromodulin in the endoplasmic reticulum (ER) and to ER hyperplasia. Further, this study was able to demonstrate for the first time in vivo that the severity of the uromodulin maturation defect as well as onset and speed of progression of renal dysfunction and morphological alterations are strongly dependent on the particular Umod mutation itself and the zygosity status.
Collapse
|
37
|
Hato T, El-Achkar TM, Dagher PC. Sisters in arms: myeloid and tubular epithelial cells shape renal innate immunity. Am J Physiol Renal Physiol 2013; 304:F1243-51. [PMID: 23515715 DOI: 10.1152/ajprenal.00101.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The importance of innate immunity for survival is underscored by its presence at almost every level of the evolutionary tree of life. The task of "danger" recognition by the innate immune system is carried out by a broad class of pattern recognition receptors. These receptors are expressed in both hematopoietic and nonhematopoietic cells such as renal epithelial cells. Upon activation, pattern recognition receptors induce essentially two types of defensive responses: inflammation and phagocytosis. In this review, we highlight evidence that renal epithelial cells are endowed with such defensive capabilities and as such fully participate in renal innate immune responses.
Collapse
Affiliation(s)
- Takashi Hato
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | | | | |
Collapse
|